Epigenetic processes participate in cancer development and likely influence cancer prevention. Global DNA hypomethylation, gene promoter hypermethylation and aberrant histone post-translational modifications are hallmarks of neoplastic cells which have been associated with genomic instability and altered gene expression. Because epigenetic deregulation occurs early in carcinogenesis and is potentially reversible, intervention strategies targeting the epigenome have been proposed for cancer prevention. Bioactive food components (BFCs) with anticancer potential, including folate, polyphenols, selenium, retinoids, fatty acids, isothiocyanates and allyl compounds, influence DNA methylation and histone modification processes. Such activities have been shown to affect the expression of genes involved in cell proliferation, death and differentiation that are frequently altered in cancer. Although the epigenome represents a promising target for cancer prevention with BFCs, few studies have addressed the influence of dietary components on these mechanisms in vivo, particularly on the phenotype of humans, and thus the exact mechanisms whereby diet mediates an effect on cancer prevention remains unclear. Primary factors that should be elucidated include the effective doses and dose timing of BFCs to attain epigenetic effects. Because diet-epigenome interactions are likely to occur in utero, the impact of early-life nutrition on cancer risk programming should be further investigated.

1.
Thun MJ, DeLancey JO, Center MM, Jemal A, Ward EM: The global burden of cancer: priorities for prevention. Carcinogenesis 2010;31:100–110.
2.
World Cancer Research Fund/American Institute for Cancer Research. Food, Nutrition, Physical Activity, and the Prevention of Cancer: A Global Perspective. Washington, American Institute for Cancer Research, 2007.
3.
Milner JA: Molecular targets for bioactive food components. J Nutr 2004;134:2492S–2498S.
4.
Surh YJ: Cancer chemoprevention with dietary phytochemicals. Nat Rev Cancer 2003;3:768–780.
5.
Davis CD, Milner JA: Molecular targets for nutritional preemption of cancer. Curr Cancer Drug Targets 2007;7:410–415.
6.
Ross SA: Diet and DNA methylation interactions in cancer prevention. Ann NY Acad Sci 2003;983:197–207.
7.
Davis CD, Uthus EO: DNA methylation, cancer susceptibility and nutrient interactions. Exp Biol Med 2004;229:988–995.
8.
Dashwood RH, Myzak MC, Ho E: Dietary HDAC inhibitors: time to rethink weak ligands in cancer chemoprevention? Carcinogenesis 2006;27:344–349.
9.
Link A, Balaguer F, Goel A: Cancer chemoprevention by dietary polyphenols: promising role for epigenetics. Biochem Pharmacol 2010;80:1771–1792.
10.
Chen J, Xu X: Diet, epigenetic, and cancer prevention. Adv Genet 2010;71:237–255.
11.
Callinan PA, Feinberg AP: The emerging science of epigenomics. Hum Mol Genet 2006;15:R95–R101.
12.
Jones PA, Baylin SB: The epigenomics of cancer. Cell 2007;128:683–692.
13.
Portela A, Esteller M: Epigenetic modifications and human disease. Nat Biotechnol 2010;28:1057–1068.
14.
Fazzari MJ, Greally JM: Introduction to epigenomics and epigenome-wide analysis. Methods Mol Biol 2010;620:243–265.
15.
Myzak MC, Dashwood RH: Histone deacetylases as targets for dietary cancer preventive agents: lessons learned with butyrate, diallyl disulfide, and sulforaphane. Curr Drug Targets 2006;7:443–452.
16.
Ferguson LR, Tatham AL, Lin Z, Denny WA: Epigenetic regulation of gene expression as an anticancer drug target. Curr Cancer Drug Targets 2011;11:199–212.
17.
Huang J, Plass C, Gerhäuser C: Cancer chemoprevention by targeting the epigenome. Curr Drug Targets 2010, E-pub ahead of print.
18.
Hilakivi-Clarke L, de Assis S: Fetal origins of breast cancer. Trends Endocrinol Metab 2006;17:340–348.
19.
Thornburg KL, Shannon J, Thuillier P, Turker MS: In utero life and epigenetic predisposition for disease. Adv Genet 2010;71:57–78.
20.
Grønbaek K, Treppendahl M, Asmar F, Guldberg P: Epigenetic changes in cancer as potential targets for prophylaxis and maintenance therapy. Basic Clin Pharmacol Toxicol 2008;103:389–396.
21.
Ehrlich M: DNA hypomethylation in cancer cells. Epigenomics 2009;1:239–259.
22.
Pogribny IP, Beland FA: DNA hypomethylation in the origin and pathogenesis of human diseases. Cell Mol Life Sci 2009;66:2249–2261.
23.
Baylin SB, Ohm JE: Epigenetic gene silencing in cancer – a mechanism for early oncogenic pathway addiction? Nat Rev Cancer 2006;6:107–116.
24.
Das PM, Singal R: DNA methylation and Cancer. J Clin Oncol 2004;22:4632–4642.
25.
Verma M, Srivastava S: Epigenetics in cancer: implications for early detection and prevention. Lancet Oncol 2002;3:755–763.
26.
Yoo CB, Jones PA: Epigenetic therapy of cancer: past, present and future. Nat Rev Drug Discov 2006;5:37–50.
27.
Füllgrabe J, Kavanagh E, Joseph B: Histone onco-modifications. Oncogene 2011;30:3391–3403.
28.
Chi P, Allis CD, Wang GG: Covalent histone modifications – miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer 2010;10:457–469.
29.
Fraga MF, Ballestar E, Villar-Garea A, Boix-Chornet M, Espada J, Schotta G, Bonaldi T, Haydon C, Ropero S, Petrie K, Iyer NG, Pérez-Rosado A, Calvo E, Lopez JA, Cano A, Calasanz MJ, Colomer D, Piris MA, Ahn N, Imhof A, Caldas C, Jenuwein T, Esteller M: Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat Genet 2005;37:391–400.
30.
Fraga MF, Esteller M: Towards the human cancer epigenome: a first draft of histone modifications. Cell Cycle 2005;4:1377–1381.
31.
Lin YW, Chen HM, Fang JY: Gene silencing by the polycomb group proteins and associations with cancer. Cancer Invest 2011;29:187–195.
32.
Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R: FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 2007;12:1247–1252.
33.
Varier RA, Timmers HTM: Histone lysine methylation and demethylation pathways in cancer. Biochim Biophys Acta 2011;1815:75–89.
34.
Davis CD, Ross SA: Dietary components impact histone modifications and cancer risk. Nutr Rev 2007;65:88–94.
35.
Rogers AE: Methyl donors in the diet and responses to chemical carcinogens. Am J Clin Nutr 1995;61:659S–665S.
36.
Poirier LA: Methyl group deficiency in hepatocarcinogenesis. Drug Metab Rev 1994;26:185–199.
37.
Pogribny IP, James SJ: De novo methylation of the p16INK4A gene in early preneoplastic liver and tumors induced by folate/methyl deficiency in rats. Cancer Lett 2002;187:69–75.
38.
Pogribny IP, Ross SA, Wise C, Pogribna M, Jones EA, Tryndyak VP, James SJ, Dragan YP, Poirier LA: Irreversible global DNA hypomethylation as a key step in hepatocarcinogenesis induced by dietary methyl deficiency. Mutat Res 2006;593:80–87.
39.
Ghoshal K, Li X, Datta J, Bai S, Pogribny I, Pogribny M, Huang Y, Young D, Jacob ST: A folate- and methyl-deficient diet alters the expression of DNA methyltransferases and methyl CpG binding proteins involved in epigenetic gene silencing in livers of F344 rats. J Nutr 2006;136:1522–1527.
40.
James SJ, Pogribny IP, Pogribna M, Miller BJ, Jernigan S, Melnyk S: Mechanisms of DNA damage, DNA hypomethylation, and tumor progression in the folate/methyl-diet rat model of hepatocarcinogenesis. J Nutr 2003;133:3740S–3747S.
41.
Pascale RM, Simile MM, De Miglio MR, Feo F: Chemoprevention of hepatocarcinogenesis: S-adenosyl-L-methionine. Alcohol 2002;27:193–198.
42.
Chagas CE, Bassoli BK, de Souza CA, Deminice R, Júnior AA, Paiva SA, Dagli ML, Ong TP, Moreno FS: Folic acid supplementation during early hepatocarcinogenesis: cellular and molecular effects. Int J Cancer 2011, E-pub ahead of print, DOI: 10.1002/ijc.25886.
43.
Tao L, Yang S, Xie M, Kramer PM, Pereira MA: Effect of trichloroethylene and its metabolites, dichloroacetic acid and trichloroacetic acid, on the methylation and expression of c-jun and c-myc protooncogenes in mouse liver: prevention by methionine. Toxicol Sci 2000;54:399–407.
44.
Duthie SJ: Folate and cancer: how DNA damage, repair and methylation impact on colon carcinogenesis. J Inherit Metab Dis 2011;34:101–109.
45.
Kim Y-In: Folate and colorectal cancer: an evidence-based critical review. Mol Nutr Food Res 2007;51:267–292.
46.
Sauer J, Mason JB, Choi SW: Too much folate: a risk factor for cancer and cardiovascular disease? Curr Opin Clin Nutr Metab Care 2009;12:30–36.
47.
Stover PJ: One-carbon metabolism-genome interactions in folate-associated pathologies. J Nutr 2009;139:2402–2405.
48.
Stidley CA, Picchi MA, Leng S, Willink R, Crowell RE, Flores KG, Kang H, Byers T, Gilliland FD, Belinsky SA: Multivitamins, folate, and green vegetables protect against gene promoter methylation in the aerodigestive tract of smokers. Cancer Res 2010;70:568–574.
49.
Wallace K, Grau MV, Levine AJ, Shen L, Hamdan R, Chen X, Gui J, Haile RW, Barry EL, Ahnen D, McKeown-Eyssen G, Baron JA, Issa JP: Association between folate levels and CpG island hypermethylation in normal colorectal mucosa. Cancer Prev Res 2010;3:1552–1564.
50.
Fang M, Chen D, Yang C: Dietary polyphenols may affect DNA methylation. J Nutr 2007;137:223S–228S.
51.
Lee W, Shim J, Zhu B: Mechanisms for the inhibition of DNA methyltransferases by tea catechins and bioflavonoids. Mol Pharmacol 2005;68:1018–1030.
52.
Berletch JB, Liu C, Love WK, Andrews LG, Katiyar SK, Tollefsbol TO: Epigenetic and genetic mechanisms contribute to telomerase inhibition by EGCG. J Cell Biochem 2008;103:509–519.
53.
Chuang JC, Yoo CB, Kwan JM, Li TW, Liang G, Yang AS, Jones PA: Comparison of biological effects of non-nucleoside DNA methylation inhibitors versus 5-aza-2′-deoxycytidine. Mol Cancer Ther 2005;4:1515–1520.
54.
Morey Kinney SR, Zhang W, Pascual M, Greally JM, Gillard BM, Karasik E, Foster BA, Karpf AR: Lack of evidence for green tea polyphenols as DNA methylation inhibitors in murine prostate. Cancer Prev Res 2009;2:1065–1075.
55.
Volate SR, Muga SJ, Issa AY, Nitcheva D, Smith T, Wargovich MJ: Epigenetic modulation of the retinoid X receptor alpha by green tea in the azoxymethane-Apc Min/+ mouse model of intestinal cancer. Mol Carcinog 2009;48:920–933.
56.
Mittal A, Piyathilake C, Hara Y, Katiyar SK: Exceptionally high protection of photocarcinogenesis by topical application of (–)-epigallocatechin-3-gallate in hydrophilic cream in SKH-1 hairless mouse model: relationship to inhibition of UVB-induced global DNA hypomethylation. Neoplasia 2003;5:555–565.
57.
Tsao AS, Liu D, Martin J, Tang XM, Lee JJ, El-Naggar AK, Wistuba I, Culotta KS, Mao L, Gillenwater A, Sagesaka YM, Hong WK, Papadimitrakopoulou V: Phase II randomized, placebo-controlled trial of green tea extract in patients with high-risk oral premalignant lesions. Cancer Prev Res 2009;2:931–941.
58.
Yuasa Y, Nagasaki H, Akiyama Y, Hashimoto Y, Takizawa T, Kojima K, Kawano T, Sugihara K, Imai K, Nakachi K: DNA methylation status is inversely correlated with green tea intake and physical activity in gastric cancer patients. Int J Cancer 2009;124:2677–2682.
59.
Fang M, Chen D, Sun Y, Jin Z, Christman J, Yang C: Reversal of hypermethylation and reactivation of p16INK4a, RAR β, and MGMT genes by genistein and other isoflavones from soy. Clin Cancer Res 2005;11:7033–7041.
60.
Majid S, Dar AA, Ahmad AE, Hirata H, Kawakami K, Shahryari V, Saini S, Tanaka Y, Dahiya AV, Khatri G, Dahiya R: BTG3 tumor suppressor gene promoter demethylation, histone modification and cell cycle arrest by genistein in renal cancer. Carcinogenesis 2009;30:662–670.
61.
Day JK, Bauer AM, DesBordes C, Zhuang Y, Kim BE, Newton LG, Nehra V, Forsee KM, MacDonald RS, Besch-Williford C, Huang TH, Lubahn DB: Genistein alters methylation patterns in mice. J Nutr 2002;132:2419S–2423S.
62.
Qin W, Zhu W, Shi H, Hewett JE, Ruhlen RL, MacDonald RS, Rottinghaus GE, Chen YC, Sauter ER: Soy isoflavones have an antiestrogenic effect and alter mammary promoter hypermethylation in healthy premenopausal women. Nutr Cancer 2009;61:238–244.
63.
Liu Z, Xie Z, Jones W, Pavlovicz RE, Liu S, Yu J, Li PK, Lin J, Fuchs JR, Marcucci G, Li C, Chan KK: Curcumin is a potent DNA hypomethylation agent. Bioorg Med Chem Lett 2009;19:706–709.
64.
Papoutsis AJ, Lamore SD, Wondrak GT, Selmin OI, Romagnolo DF: Resveratrol prevents epigenetic silencing of BRCA-1 by the aromatic hydrocarbon receptor in human breast cancer cells. J Nutr 2010;140:1607–1614.
65.
Lee WJ, Zhu BT: Inhibition of DNA methylation by caffeic acid and chlorogenic acid, two common catechol-containing coffee polyphenols. Carcinogenesis 2006;27:269–277.
66.
Xiang N, Zhao R, Song G, Zhong W: Selenite reactivates silenced genes by modifying DNA methylation and histones in prostate cancer cells. Carcinogenesis 2008;29:2175–2181.
67.
Davis CD, Uthus EO, Finley JW: Dietary selenium and arsenic affect DNA methylation in vitro in Caco-2 cells and in vivo in rat liver and colon. J Nutr 2000;130:2903–2909.
68.
Davis CD, Uthus EO: Dietary selenite and azadeoxycytidine treatments affect dimethylhydrazine-induced aberrant crypt formation in rat colon and DNA methylation in HT-29 cells. J Nutr 2002;132:292–297.
69.
Uthus EO, Ross SA, Davis CD: Differential effects of dietary selenium (Se) and folate on methyl metabolism in liver and colon of rats. Biol Trace Elem Res 2006;109:201–214.
70.
Ramachandran K, Navarro L, Gordian E, Das PM, Singal R: Methylation-mediated silencing of genes is not altered by selenium treatment of prostate cancer cells. Anticancer Res 2007;27:921–925.
71.
Pilsner JR, Hall MN, Liu X, Ahsan H, Ilievski V, Slavkovich V, Levy D, Factor-Litvak P, Graziano JH, Gamble MV: Associations of plasma selenium with arsenic and genomic methylation of leukocyte DNA in Bangladesh. Environ Health Perspect 2011;119:113–118.
72.
Johanning GL, Piyathilake CJ: Retinoids and epigenetic silencing in cancer. Nutr Rev 2003;61:284–289.
73.
Rowling MJ, McMullen MH, Schalinske KL: Vitamin A and its derivatives induce hepatic glycine N-methyltransferase and hypomethylation of DNA in rats. J Nutr 2002;132:365–369.
74.
Moreno FS, S-Wu T, Naves MM, Silveira ER, Oloris SC, da Costa MA, Dagli ML, Ong TP: Inhibitory effects of beta-carotene and vitamin A during the progression phase of hepatocarcinogenesis involve inhibition of cell proliferation but not alterations in DNA methylation. Nutr Cancer 2002;44:80–88.
75.
Esteller M, Guo M, Moreno V, Peinado MA, Capella G, Galm O, Baylin SB, Herman JG: Hypermethylation-associated inactivation of the cellular retinol-binding-protein 1 gene in human cancer. Cancer Res 2002;62:5902–5905.
76.
Di Croce L, Raker VA, Corsaro M, Fazi F, Fanelli M, Faretta M, Fuks F, Lo Coco F, Kouzarides T, Nervi C, Minucci S, Pelicci PG: Methyltransferase recruitment and DNA hypermethylation of target promoters by an oncogenic transcription factor. Science 2002;295:1079–1082.
77.
Fazi F, Travaglini L, Carotti D, Palitti F, Diverio D, Alcalay M, McNamara S, Miller WH Jr, Lo Coco F, Pelicci PG, Nervi C: Retinoic acid targets DNA-methyltransferases and histone deacetylases during APL blast differentiation in vitro and in vivo. Oncogene 2005;24:1820–1830.
78.
Stefanska B, Rudnicka K, Bednarek A, Fabianowska-Majewska K: Hypomethylation and induction of retinoic acid receptor beta 2 by concurrent action of adenosine analogues and natural compounds in breast cancer cells. Eur J Pharmacol 2010;638:47–53.
79.
Cheong HS, Lee HC, Park BL, Kim H, Jang MJ, Han YM, Kim SY, Kim YS, Shin HD: Epigenetic modification of retinoic acid-treated human embryonic stem cells. BMB Rep 2010;43:830–835.
80.
Chung TL, Brena RM, Kolle G, Grimmond SM, Berman BP, Laird PW, Pera MF, Wolvetang EJ: Vitamin C promotes widespread yet specific DNA demethylation of the epigenome in human embryonic stem cells. Stem Cells 2010;28:1848–1855.
81.
Nian H, Delage B, Ho E, Dashwood RH: Modulation of histone deacetylase activity by dietary isothiocyanates and allyl sulfides: studies with sulforaphane and garlic organosulfur compounds. Environ Mol Mutagen 2009;50:213–221.
82.
Lee JI, Nian H, Cooper AJ, Sinha R, Dai J, Bisson WH, Dashwood RH, Pinto JT: Alpha-keto acid metabolites of naturally occurring organoselenium compounds as inhibitors of histone deacetylase in human prostate cancer cells. Cancer Prev Res 2009;2:683–693.
83.
Kashyap V, Gudas LJ: Epigenetic regulatory mechanisms distinguish retinoic acid-mediated transcriptional responses in stem cells and fibroblasts. J Biol Chem 2010;285:14534–14548.
84.
Dimri M, Bommi PV, Sahasrabuddhe AA, Khandekar JD, Dimri GP: Dietary omega-3 polyunsaturated fatty acids suppress expression of EZH2 in breast cancer cells. Carcinogenesis 2010;31:489–495.
85.
Chen YX, Fang JY, Zhu HY, Lu R, Cheng ZH, Qiu DK: Histone acetylation regulates p21WAF1 expression in human colon cancer cell lines. World J Gastroenterol 2004;10:2643–2646.
86.
Davie JR: Inhibition of histone deacetylase activity by butyrate. J Nutr 2003;133:2485S–2493S.
87.
Waby JS, Chirakkal H, Yu C, Griffiths GJ, Benson RS, Bingle CD, Corfe BM: Sp1 acetylation is associated with loss of DNA binding at promoters associated with cell cycle arrest and cell death in a colon cell line. Mol Cancer 2010;9:275.
88.
Shen WJ, Dai DQ, Teng Y, Liu HB: Regulation of demethylation and re-expression of RASSF1A gene in gastric cancer cell lines by combined treatment of 5-Aza-CdR and NaB. World J Gastroenterol 2008;14:595–600.
89.
De Conti, Kuroiwa-Trzmielina J, Horst MA, Bassoli BK, Chagas CE, Purgatto E, Cavalher FP, Camargo AA, Jordão Junior AA, Vannucchi H, Scolastici C, Ong TP, Moreno FS: Chemopreventive effects of the dietary histone deacetylase inhibitor tributyrin alone or in combination with vitamin A during the promotion phase of rat hepatocarcinogenesis. J Nutr Biochem, 2011, E-pub ahead of print.
90.
Lu R, Wang X, Sun DF, Tian XQ, Zhao SL, Chen YX, Fang JY: Folic acid and sodium butyrate prevent tumorigenesis in a mouse model of colorectal cancer. Epigenetics 2008;3:330–335.
91.
Kuroiwa-Trzmielina J, de Conti A, Scolastici C, Pereira D, Horst MA, Purgatto E, Ong TP, Moreno FS: Chemoprevention of rat hepatocarcinogenesis with histone deacetylase inhibitors: efficacy of tributyrin, a butyric acid prodrug. Int J Cancer 2009;124:2520–2527.
92.
Myzak MC, Karplus PA, Chung FL, Dashwood RH: A novel mechanism of chemoprotection by sulforaphane: inhibition of histone deacetylase. Cancer Res 2004;64:5767–5774.
93.
Myzak MC, Hardin K, Wang R, Dashwood RH, Ho E: Sulforaphane inhibits histone deacetylase activity in BPH-1, LnCaP and PC-3 prostate epithelial cells. Carcinogenesis 2006;27:811–819.
94.
Hu R, Khor TO, Shen G, Jeong WS, Hebber V, Chen C, Xu C, Reddy B, Chada K, Kong AN: Cancer chemoprevention of intestinal polyposis in ApcMin/+ mice by sulforaphane, a natural product derived from cruciferous vegetables. Carcinogenesis 2006;27:2038–2046.
95.
Clarke JD, Hsu A, Yu Z, Dashwood RH, Ho E: Differential effects of sulforaphane on histone deacetylases, cell cycle arrest and apoptosis in normal prostate cells versus hyperplastic and cancerous prostate cells. Mol Nutr Food Res 2011;55:999–1009.
96.
Pledgie-Tracy A, Sobolewski MD, Davidson NE: Sulforaphane induces cell type-specific apoptosis in human breast cancer cell lines. Mol Cancer Ther 2007;6:1013–1021.
97.
Myzak MC, Dashwood WM, Orner GA, Ho E, Dashwood RH: Sulforaphane inhibits histone deacetylase in vivo and suppresses tumorigenesis in Apc-minus mice. FASEB J 2006;20:506–508.
98.
Myzak MC, Tong P, Dashwood WM, Dashwood RH, Ho E: Sulforaphane retards the growth of human PC-3 xenografts and inhibits HDAC activity in human subjects. Exp Biol Med 2007;232:227–234.
99.
Ma X, Fang Y, Beklemisheva A, Dai W, Feng J, Ahmed T, Liu D, Chiao JW: Phenylhexyl isothiocyanate inhibits histone deacetylases and remodels chromatins to induce growth arrest in human leukemia cells. Int J Oncol 2006;28:1287–1293.
100.
Wang LG, Liu XM, Fang Y, Dai W, Chiao FB, Puccio GM, Feng J, Liu D, Chiao JW: De-repression of the p21 promoter in prostate cancer cells by an isothiocyanate via inhibition of HDACs and c-Myc. Int J Oncol 2008;33:375–380.
101.
Lea MA, Randolph VM, Patel M: Increased acetylation of histones induced by diallyl disulfide and structurally related molecules. Int J Oncol 1999;15:347–352.
102.
Lea MA, Randolph VM: Induction of histone acetylation in rat liver and hepatoma by organosulfur compounds including diallyl disulfide. Anticancer Res 2001;21:2841–2845.
103.
Druesne N, Pagniez A, Mayeur C, Thomas M, Cherbuy C, Duée PH, Martel P, Chaumontet C: Diallyl disulfide (DADS) increases histone acetylation and p21(waf1/cip1) expression in human colon tumor cell lines. Carcinogenesis 2004;25:1227–1236.
104.
Druesne-Pecollo N, Pagniez A, Thomas M, Cherbuy C, Duée PH, Martel P, Chaumontet C: Diallyl disulfide increases CDKN1A promoter-associated histone acetylation in human colon tumor cell lines. J Agric Food Chem 2006;54:7503–7507.
105.
Zhao J, Huang WG, He J, Tan H, Liao QJ, Su Q: Diallyl disulfide suppresses growth of HL-60 cell through increasing histone acetylation and p21WAF1 expression in vivo and in vitro. Acta Pharmacol Sin 2006;27:1459–1466.
106.
Nian H, Delage B, Pinto JT, Dashwood RH: Allyl mercaptan, a garlic-derived organosulfur compound, inhibits histone deacetylase and enhances Sp3 binding on the P21WAF1 promoter. Carcinogenesis 2008;29:1816–1824.
107.
Kikuno N, Shiina H, Urakami S, Kawamoto K, Hirata H, Tanaka Y, Majid S, Igawa M, Dahiya R: Genistein mediated histone acetylation and demethylation activates tumor suppressor genes in prostate cancer cells. Int J Cancer 2008;123:552–560.
108.
Majid S, Kikuno N, Nelles J, Noonan E, Tanaka Y, Kawamoto K, Hirata H, Li LC, Zhao H, Okino ST, Place RF, Pookot D, Dahiya R: Genistein induces the p21WAF1/CIP1 and p16INK4a tumor suppressor genes in prostate cancer cells by epigenetic mechanisms involving active chromatin modification. Cancer Res 2008;68:2736–2744.
109.
Li Y, Liu L, Andrews LG, Tollefsbol TO: Genistein depletes telomerase activity through cross-talk between genetic and epigenetic mechanisms. Int J Cancer 2009;125:286–296.
110.
Kang SK, Cha SH, Jeon HG: Curcumin-induced histone hypoacetylation enhances caspase-3-dependent glioma cell death and neurogenesis of neural progenitor cells. Stem Cells Dev 2006;15:165–174.
111.
Rajendran P, Williams DE, Ho E, Dashwood RH: Metabolism as a key to histone deacetylase inhibition. Crit Rev Biochem Mol Biol 2011;46:181–199.
112.
Lee JI, Nian H, Cooper AJ, Sinha R, Dai J, Bisson WH, Dashwood RH, Pinto JT: Alpha-keto acid metabolites of naturally occurring organoselenium compounds as inhibitors of histone deacetylase in human prostate cancer cells. Cancer Prev Res 2009;2:683–693.
113.
Bracken AP, Helin K: Polycomb group proteins: navigators of lineage pathways led astray in cancer. Nat Rev Cancer 2009;9:773–784.
114.
Prezioso C, Orlando V: Polycomb proteins in mammalian cell differentiation and plasticity. FEBS Lett 2011;585:2067–2077.
115.
Piunti A, Pasini D: Epigenetic factors in cancer development: polycomb group proteins. Future Oncol 2011;7:57–75.
116.
Gudas LJ, Wagner JA: Retinoids regulate stem cell differentiation. J Cell Physiol 2011;226:322–330.
117.
Lee K, Adhikary G, Balasubramanian S, Gopalakrishna R, McCormick T, Dimri GP, Eckert RL, Rorke EA: Expression of Bmi-1 in epidermis enhances cell survival by altering cell cycle regulatory protein expression and inhibiting apoptosis. J Invest Dermatol 2008;128:9–17.
118.
Balasubramanian S, Lee K, Adhikary G, Gopalakrishnan R, Rorke EA, Eckert RL: The Bmi-1 polycomb group gene in skin cancer: regulation of function by (–)-epigallocatechin-3-gallate (EGCG). Nutr Rev 2008;66:S65–S68.
119.
Balasubramanian S, Adhikary G, Eckert RL: The Bmi-1 polycomb protein antagonizes the (–)-epigallocatechin-3-gallate-dependent suppression of skin cancer cell survival. Carcinogenesis 2010;31:496–503.
120.
Hua WF, Fu YS, Liao YJ, Xia WJ, Chen YC, Zeng YX, Kung HF, Xie D: Curcumin induces down-regulation of EZH2 expression through the MAPK pathway in MDA-MB-435 human breast cancer cells. Eur J Pharmacol 2010;637:16–21.
121.
Widshwendter M, Fiegl H, Egle D, Mueller-Holzner E, Spizzo G, Marth C, Weisenberger DJ, Campan M, Young J, Jacobs I, Laird PW: Epigenetic stem cell signature in cancer. Nat Genet 2007;39:157–158.
122.
Wells PG, McCallum GP, Chen CS, Henderson JT, Lee CJ, Perstin J, Preston TJ, Wiley MJ, Wong AW: Oxidative stress in developmental origins of disease: teratogenesis, neurodevelopmental deficits, and cancer. Toxicol Sci 2009;108: 4–18.
123.
Perera F: Molecular epidemiology, prenatal exposure and prevention of cancer. Environ Health 2011;10:S5.
124.
Vessey MP: Epidemiological studies of the effects of diethylstilboestrol. IARC Sci Publ 1989;96:335–348.
125.
Soto AM, Vandenberg LN, Maffini MV, Sonnenschein C: Does breast cancer start in the womb? Basic Clin Pharmacol Toxicol 2008;102:125–133.
126.
Trichopoulos D: Hypothesis: does breast cancer originate in utero? Lancet 1990;335:939–940.
127.
Hilakivi-Clarke L: Nutritional modulation of terminal end buds: its relevance to breast cancer prevention. Curr Cancer Drug Targets 2007;7:465–474.
128.
Nafee T, Farrell W, Carroll W, Fryer A, Ismail K: Epigenetic control of fetal gene expression. BJOG 2008;115:158–168.
129.
Attig L, Gabory A, Junien C: Nutritional developmental epigenomics: immediate and long-lasting effects. Proc Nutr Soc 2010;69:221–231.
130.
Chiam K, Tilley WD, Butler LM, Bianco-Miotto T: The dynamic and static modification of the epigenome by hormones: a role in the developmental origin of hormone related cancers. Biochim Biophys Acta 2009;1795:104–109.
131.
Yenbutr P, Hilakivi-Clarke L, Passaniti A: Hypomethylation of an exon I estrogen receptor CpG island in spontaneous and carcinogen-induced mammary tumorigenesis in the rat. Mech Ageing Dev 1998;106:93–102.
132.
Waterland RA, Jirtle RL: Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol Cell Biol 2003;23:5293–5300.
133.
Dolinoy DC, Weidman JR, Waterland RA, Jirtle RL: Maternal genistein alters coat color and protects Avy mouse offspring from obesity by modifying the fetal epigenome. Environ Health Perspect 2006;114:567–572.
134.
Lillycrop KA, Phillips ES, Jackson AA, Hanson MA, Burdge GC: Dietary protein restriction of pregnant rats induces and folic acid supplementation prevents epigenetic modification of hepatic gene expression in the offspring. J Nutr 2005;135:1382–1386.
135.
Lillycrop KA, Slater-Jefferies JL, Hanson MA, Godfrey KM, Jackson AA, Burdge GC: Induction of altered epigenetic regulation of the hepatic glucocorticoid receptor in the offspring of rats fed a protein-restricted diet during pregnancy suggests that reduced DNA methyltransferase-1 expression is involved in impaired DNA methylation and changes in histone modifications. Br J Nutr 2007;97:1064–1073.
136.
Aagaard-Tillery KM, Grove K, Bishop J, Ke X, Fu Q, McKnight R, Lane RH: Developmental origins of disease and determinants of chromatin structure: maternal diet modifies the primate fetal epigenome. Mol Endocrinol 2008;41:91–102.
137.
Foley DL, Craig JM, Morley R, Olsson CA, Dwyer T, Smith K, Saffery R: Prospects for epigenetic epidemiology. Am J Epidemiol 2009;169:389–400.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.
You do not currently have access to this content.